Critical Insights into Cardiovascular Disease from Basic Research on

Aug 26, 2011 - polyunsaturated phospholipids, is delivering a bonanza of molecular mechanistic insights into cardiovascular disease. Rather than targe...
0 downloads 0 Views
REVIEW pubs.acs.org/crt

Critical Insights into Cardiovascular Disease from Basic Research on the Oxidation of Phospholipids: The γ-Hydroxyalkenal Phospholipid Hypothesis Robert G. Salomon*,† and Xiaodong Gu†,‡ † ‡

Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106-7078, United States Department of Cell Biology, Cleveland Clinic Foundation, Cleveland Ohio 44195, United States ABSTRACT:

Basic research, exploring the hypothesis that γ-hydroxyalkenal phospholipids are generated in vivo through oxidative cleavage of polyunsaturated phospholipids, is delivering a bonanza of molecular mechanistic insights into cardiovascular disease. Rather than targeting a specific pathology, these studies were predicated on the presumption that a fundamental understanding of lipid oxidation is likely to provide critical insights into disease processes. This investigational approach, from the chemistry of biomolecules to disease phenotype, that complements the more common opposite paradigm, is proving remarkably productive.

’ CONTENTS Introduction 1792 γ-Hydroxyalkenal Phospholipids: From the Hypothesis to the Detection of Protein Adducted Derivatives in Vivo 1792 γ-Hydroxyalkenal Phospholipid Hypothesis Immunological Detection of Cardiovascular Disease-Related Levels of γ-Hydroxyalkenal Phospholipid-Derived Protein Adducts in Vivo γ-Hydroxyalkenal Phospholipids: From Synthesis to Detection in Vivo and the Discovery of Biological Activities LC-MS/MS Characterization of Biologically Active Oxidized Lipids in Complex Mixtures HOOA-PC Promotes Monocyte Entry into Chronic Lesions Lipid Whisker Model for Membrane Phospholipids γ-Hydroxyalkenal Phospholipids and Their More Oxidized Derivatives Are Scavenger Receptor CD36 Ligands OxPCCD36 Accumulate in Atherosclerotic Lesions and Foster Foam Cell Formation OxLDL Is a Trojan Horse That Delivers Toxic Electropohilic γ-Hydroxyalkenals into Macrophage and RPE Cells r 2011 American Chemical Society

1792

1792

1793 1793 1793 1793

1794

OxPCCD36 Inhibit HDL Binding with Hepatocyte SRB1, Impeding the Delivery of Cholesterol to the Liver for Excretion OxPCCD36 Induce a Prothrombotic State through the Activation of Platelet CD36 Oxidatively Truncated Ether PCs Activate Platelets through the PAF Receptor α,β-Unsaturated Carboxylic oxPEs and oxPCs Inhibit LPS-Induced Expression of IL-8 γ-Hydroxyalkenal Phospholipids: Biologically Important Chemistry Membrane Lipid Composition Profoundly Influences the Stability of γ-Hydroxy-α,β-unsaturated Aldehydic PCs Spontaneous Cyclodehydration of HODA-PCs Abolishes Recognition by the Scavenger Receptor CD36 Caveat: Lyso-PC Is Generated by Spontaneous Nonenzymatic Deacylation of oxPCs Conclusions Author Information Acknowledgment Abbreviations References

1796 1796 1797 1797 1798

1798

1798 1799 1800 1800 1800 1800 1801

1795

1796

Received: May 15, 2011 Published: August 26, 2011 1791

dx.doi.org/10.1021/tx200207z | Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

’ INTRODUCTION The premise that a fundamental understanding of phospholipid oxidation1 11 is likely to provide critical insights into disease processes, e.g., atherogenesis,12 15 led us to predict and explore the possibility that γ-hydroxyalkenal phospholipids are generated in vivo. The resulting studies have now delivered a bonanza of critical molecular mechanistic insights into the pathogenesis of cardiovascular disease. A key feature of those studies is the chemical synthesis of pure samples of putative natural products16 that facilitated the development of analytical methods for detecting and quantifying their natural occurrence, exploring their biologically important chemistry, and assessing their biological activities. This investigational approach, from biomolecular chemistry to the disease phenotype, that complements the more common opposite paradigm is proving to be remarkably productive. This review focuses on γ-hydroxyalkenal phospholipids, especially their contributions to the pathogenesis of cardiovascular disease. A companion review describes how the covalent modification of proteins by γ-hydroxyalkenal phospholipids, to generate carboxyalkyl pyrrole derivatives, contributes to age-related macular degeneration, autism, cancer, and wound healing.17 ’ γ-HYDROXYALKENAL PHOSPHOLIPIDS: FROM THE HYPOTHESIS TO THE DETECTION OF PROTEIN ADDUCTED DERIVATIVES IN VIVO γ-Hydroxyalkenal Phospholipid Hypothesis. Oxidative cleavage of bond “a” of 1-palmityl-2-arachidonyl-sn-glycero-3-

Figure 1. Free radical-induced oxidative cleavage of PA-PC to generate HOOA-PC, a γ-hydroxyalkenal phospholipid analogue of HNE.

REVIEW

phosphocholine (PA-PC) (Figure 1) generates 1-palmityl-2-(ωoxovaleryl)-sn-glycero-3-phosphocholine (OV-PC)18 that is oxidized further to deliver 1-palmityl-2-glutaryl-sn-glycero-3-phosphocholine (G-PC). We prepared authentic samples of OV-PC and G-PC by chemical syntheses to facilitate studies demonstrating their biological activities that include promoting the adhesion of monocytes to vascular endothelial cells resulting in extravasation into the subendothelial space, an early event in the development of atherosclerosis (vide infra).19 OV-PC and G-PC are components of a complex mixture of oxidized phospholipids present in oxidatively damaged low-density lipoprotein (oxLDL). Another component of the mixture generated by oxidative cleavage of PA-PC exhibited an exact mass m/z = 650. Rather than attempting to isolate and characterize this oxidized phospholipid component of oxLDL, we opted to predict a likely candidate. By analogy with the well-known production of the γ-hydroxyalkenal 4-hydroxy-2-nonenal (HNE)20 through free radical-induced oxidative cleavage of bond “c” of PA-PC, we postulated that cleavage of bond “b” would generate a γ-hydroxyalkenal phospholipid, 1-palmityl-2-(5-hydroxy-8-oxooct-6-enoyl)-sn-glycero3-phosphocholine (HOOA-PC) that has an exact mass m/z = 650. Although free HOOA-PC would eventually be detected in vivo, we decided to test the γ-hydroxyalkenal phospholipid hypothesis through experiments that used antibodies, raised against their anticipated protein adducts, to secure the first evidence supporting their generation in vivo. Immunological Detection of Cardiovascular Disease-Related Levels of γ-Hydroxyalkenal Phospholipid-Derived Protein Adducts in Vivo. Previously, we found that HNE forms covalent adducts that incorporate the ε-amino group of protein lysyl residues in pentylpyrrole protein modifications (PPprotein in Figure 2).21 Analogous reactions of γ-hydroxyalkenal phospholipids, followed by lipolysis of intermediate phospholipid adducts, were expected to produce ω-carboxyalkylpyrrole modifications of proteins. Enzyme-linked immunosorbent assays, using antibodies raised against carboxyheptyl pyrrole (CHP) and carboxypropyl pyrrole (CPP) protein modifications (Figure 2), revealed the presence of CHPs and CPPs in oxLDL.22 Since 1-palmityl-2-linoleyl-sn-glycero-3-phosphocholine (PL-PC) is the most abundant polyunsaturated phospholipid in LDL, we also measured CHP immunoreactivity in human blood plasma. The mean level ((SD) of this protein adduct of HODA in the plasma from patients with end-stage renal disease (209 ( 32 pmol/mL, n = 16) is significantly elevated (P < 0.002) compared to that in healthy volunteers (169 ( 28 pmol/mL, n = 12). The mean level in the plasma of atherosclerosis patients

Figure 2. Covalent adduction of γ-hydroxyaldehydes with proteins generates alkyl and ω-carboxyalkyl pyrrole modifications. 1792

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

REVIEW

Figure 3. Negtative ion ESI-MS/MS of isomeric products from oxidative fragmentation of linoleate.

(209 ( 19 pmol/mL, n = 13) is also significantly elevated (P < 0.003), reminiscent of the elevated levels of other lipid-derived oxidative protein modifications associated with atherosclerosis.23 These observations supported the conclusion that HODA protein adducts are produced in vivo through covalent adduction of HODA-PC, a component of the mixture of oxidized lipids derived from PA-PC and referred to collectively as oxPA-PC. The investigation of γ-hydroxyalkenal phospholipids and the ω-carboxyalkylpyrrole derivatives produced from their adduction with proteins was not targeted at understanding the molecular basis of a particular disease. As will be described in this review and a companion review,17 nevertheless, it led to major insights into a vast array of pathological and physiological involvements of this biomolecular chemistry.

’ γ-HYDROXYALKENAL PHOSPHOLIPIDS: FROM SYNTHESIS TO DETECTION IN VIVO AND THE DISCOVERY OF BIOLOGICAL ACTIVITIES LC-MS/MS Characterization of Biologically Active Oxidized Lipids in Complex Mixtures. Free radical-induced oxida-

tion of phospholipids generates a vast array of products. The classical approach for identifying molecular structures of biologically active natural products contained in complex mixtures exploits activity assays24 to guide isolation. An alternative approach is to predict likely candidates by mechanistic speculation or analogy with known products of lipid oxidation and to use authentic samples of the putative natural products, prepared by unambiguous chemical synthesis, to guide their detection in vivo. However, the complexity of lipid extracts confounds isolation of analytically pure samples of oxidized lipids. LC-MS/MS analysis provides a workaround often allowing the detection of individual species in complex mixtures without the need for isolation. The exquisite complexity of product mixtures generated by free radicalinduced oxidation of lipids25 complicates the application of this approach because mass spectra of isobaric compounds not only exhibit identical parent ions but also may produce nearly identical fragmentation patterns and chromatographic retention times. Consequently, LC-MS/MS comparisons, even when pure authentic samples are available by chemical syntheses, must include additional chemical tests to confirm the presumed structures of components of product mixtures generated by nonenzymatic oxidation of polyunsaturated fatty acid (PUFA) derivatives. A case in point is provided

by two isomeric products generated by oxidative fragmentation of linoleic acid, the ketoaldehyde and an isobaric butenolide shown in Figure 3. Their mass spectra are nearly identical, and both compounds exhibit identical HPLC retention times with a methanol/ water solvent system. Using pure samples available by unambiguous chemical syntheses, we were able to define HPLC conditions that could distinguish them by using an acetonitrile/water solvent system. Furthermore, diagnostic derivatization clearly distinguishes the ketoaldehyde that reacts with methoxylamine (to form a bis methoxime) from the butenolide that does not. HOOA-PC Promotes Monocyte Entry into Chronic Lesions. We first executed a chemical synthesis of HOOA-PC (Figures 1 and 2)26 to facilitate its identification by LC-MS/MS comparison with components of oxPA-PC generated by the nonenzymatic oxidation of PA-PC and to enable biological testing. HOOA-PC exhibited proinflammatory activities,27 discovered previously for OV-PC and G-PC,19 which can regulate leukocyte endothelial interaction resulting in atherogenic extravasation of monocytes into the subendothelial space (Figure 4). Thus, HOOA-PC dose-dependently activates human aortic endothelial cells to bind monocytes and increases levels of monocyte chemotactic protein-1 and interleukin-8 (IL-8), chemokines that are important in monocyte entry into chronic lesions. This suggested that HOOA-PC plays a role in chronic inflammation. In a model of bacterial infection, HOOA-PC also promotes the antiinflammatory inhibition of lipopolysaccharide (LPS)-induced expression of E-Selectin, a major adhesion molecule that mediates neutrophil endothelial interactions.27 Subsequently, HOOA-PC was also found in lipid extracts from oxLDL and human atheroma (vide infra).28,29 Since elevated levels of myeloperoxidase (MPO) are associated with the presence of coronary artery disease,30 one likely scenario for atherogenesis involves MPO-initiated free radical-induced lipid oxidation (Figure 4). The resulting oxPC promote extravasation of monocyte macrophages into the subendothelial space and endocytosis of oxLDL by the macrophages leading to their conversion into foam cells and, ultimately, atheroma formation. Lipid Whisker Model for Membrane Phospholipids. Besides their utility as standards for identification and quantification of oxPC in biological samples, the availability of pure individual oxPCs through chemical syntheses also enabled a study of their conformations in membranes. Nuclear Overhauser effect experiments revealed the close proximity of oxidatively truncated sn-2 1793

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

REVIEW

Figure 4. Monocytes and endothelial cells contribute to one pathway leading to the oxidation of low-density lipoprotein (LDL) by iNOS- or eNOS-promoted generation of NO that is converted into the reactive nitrogen radical •NO2 by myeloperoxidase (MPO)-catalyzed reaction with H2O2. Low levels of oxidatively truncated phospholipids, present in minimally oxidized (mox)LDL, are sufficient to promote the adhesion of monocytes to the vascular endothelium that facilitates extravasation into the subendothelial space where they become macrophages. There, a family of γ-oxygenated-α,β-unsaturated aldehydic and carboxylic oxidatively truncated phospholipids (oxPCCD36), that protrude like whiskers from the outer shell of oxLDL, bind strongly with the scavenger receptor CD36, promoting endocytosis of oxLDL by the macrophages and leading to foam cell formation and atherogenesis. Illustration by David Schumick, BS, CMI. Reprinted with permission from Cleveland Clinic Center for Medical Art & Photography. Copyright 2011. All rights reserved.

acyl chains with the choline headgroup.31 Oxidation resulting in the sn-2 acyl chain of phospholipids becoming shorter and richer in polar functional groups increases their hydrophilicity leading to their expulsion from the hydrophobic core of the membrane lipid bilayer (Figure 5). Thus, when cellular membranes are oxidatively damaged, truncated acyl chains sprout from the membrane-like whiskers.32 Molecular dynamics simulations provide support for this lipid whisker model for oxidatively damaged membranes.33 The conformational change that results in the sprouting of lipid whiskers may have several consequences (vide infra): (a) facilitating specific binding with receptors (Figure 6), e.g., macrophage or platelet CD36 and hepatocyte SR-BI; (b) inducing higher local positive membrane curvature owing to the presence of one rather than two lipophilic tails inserted into the membrane; and (c) allowing interaction and transfer of oxPC, e.g., from oxLDL, to other lipoprotein particles through specific binding interactions, e.g., with small high-density lipoprotein (HDL) particles or peptide fragment analogues of the HDL protein Apo-A1.34 γ-Hydroxyalkenal Phospholipids and Their More Oxidized Derivatives Are Scavenger Receptor CD36 Ligands. The recognition of oxLDL by the scavenger receptor (SR) CD36 was known to trigger its endocytosis by macrophages. However, unlike the LDL receptor of macrophages that recognizes the protein component apoB100 of LDL, our collaborators at the Cleveland Clinic Foundation discovered that lipid components of oxLDL bind with CD36. To identify oxidized phosphatidylcholines (oxPCs) that bind with CD36, a simpler system, oxPA-PC was separated by HPLC to give three active fractions. Serendipitously, our chemical synthesis of HOOA-PC provided the key to rapid identification of the CD36 ligands in these mixtures. Thus, the

identification of HOOA-PC as a component of the complex mixture of oxPCs in one of the CD36 active fractions was confirmed by the identity of its LC-MS/MS retention time and fragmentation pattern with that of a pure sample, prepared by unambiguous chemical synthesis, as well as those of derivatives generated by treatment of the CD36 active fraction with multiple agents (NaBH4, NaBD4, and NaCNBH3) for reducing the aldehyde carbonyl, methoxyamine, and dinitrophenyhydrazine for generating a methoxime and hydrazone derivative and subsequent tandem MS analysis in both positive ion and negative ion modes.35 Presuming that the CD36 ligands contained in the other two active fractions from oxPA-PC were derivatives of HOOA-PC, we then postulated that a less polar fraction contained the less polar derivative KOOA-PC (Figure 7). Unambiguous chemical synthesis of KOOA-PC36 followed by LC/ ESI/MS/MS comparison with the less polar fraction and derivatives confirmed the presence of this oxPC in oxPA-PC. We similarly postulated and confirmed by chemical synthesis, derivatizations (including esterification with pentafluorobenzyl bromide), and LC/ESI/MS/MS comparisons that a more polar fraction contained a mixture of HOdiA-PC and KOdiA-PC.36 The phospholipids in oxLA-PC, which are capable of binding with CD36, were similarly identified by chemical syntheses of likely candidates and derivatizations followed by LC/ESI/MS/MS comparisons and shown to be HODA-, KODA-, HDdiA-, and KDdiA-PC (Figure 7). All of these oxPCs, which contain γ-oxygenated-α,βunsaturated aldehyde or carboxylic acid functionality, referred to collectively as oxPCCD36 (Figure 7), were confirmed to be especially potent inducers of CD36-mediated endocytosis of oxLDL by macrophage cells. 1794

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology OxPCCD36 Accumulate in Atherosclerotic Lesions and Foster Foam Cell Formation. LC/ESI/MS/MS analysis of lipid

extracts from rabbit aortas confirmed the presence of each oxPCCD36 species in vivo and demonstrated 5 7-fold elevated levels of PA-PC-derived oxPCCD36 in atherosclerotic versus normal aortas.28 Identification of each species was based upon the detection of ions with a mass-to-charge (m/z) ratio identical to that of the parent lipid, which following collision-induced dissociation, subsequently also gave rise to a characteristic daughter ion and retention time determined by an analysis of authentic synthetic oxPC species. While their presence in lipid extracts was established by LC-MS/MS comparisons with pure synthetic

Figure 5. Lipid whisker model of oxidatively damaged membranes. The more hydrophilic ω-oxovaleryl and 5-ketooct-6-endioyl-PC (KOdiAPC) but not the more lipophilic 13-hydroxyoctadeca-9,11-dienoyl-PC (13-HODE-PC) side chains protrude from the membrane where they are in close proximity with the choline headgroup and well positioned for interaction with receptors, e.g., on macrophages, platelets, or retinal pigmented epithelial (RPE) cells.

REVIEW

oxPCs and their characteristic derivatives, analytically pure samples have never been isolated from oxPA-PC, oxPL-PC, oxLDL, or human tissues. Therefore, chemical syntheses of pure samples of each oxPCCD36 were absolutely essential for the unambiguous demonstration of their biological activities. Addition of pure synthetic oxPCCD36 to cholesterol-containing particles promotes CD36-dependent macrophage binding (but not to macrophage cells from CD36 null mice), uptake, and metabolism of cholesterol esters resulting in the accumulation of cholesterol and foam cell formation.28 Notably, binding of oxPCCD36-containing particles to CD36 increased with increasing mol % of ligand within the surface of a particle, consistent with the enhancement of binding through mutivalent, i.e., multiple receptor ligand, interactions. That only a few molecules per LDL particle are needed to confer recognition of oxPCCD36 supports their physiological relevance. Analogously, oxPCCD36 promote endocytosis of oxidatively damaged photoreceptor rod cell outer segments (PhROS) by retinal pigmented epithelial (RPE) cells,37 a process that replaces the entire stack of photoreceptor disks within these cells every 10 days.

Figure 7. Oxidatively truncated phospholipids with a terminal γoxygenated α,β-unsaturated carbonyl, referred to collectively as oxPCCD36, bind avidly with the scavenger receptor CD36.

Figure 6. Macrophage scavenger receptor-CD36 detects lipid whiskers, oxidized truncated acyl chains that protrude from an oxidatively damaged cell membrane. Binding is especially strong with oxPCCD36, i.e., γ-hydroxyalkenal phospholipids and their more oxidized derivatives, that incorporate a terminal γ-oxygenated α,β-unsaturated carbonyl. Illustration by David Schumick, BS, CMI. Reprinted with permission from Cleveland Clinic Center for Medical Art & Photography. Copyright 2011. All rights reserved. 1795

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

REVIEW

Figure 8. Binding of HDL to hepatocyte SR-B1, which promotes reverse cholesterol transport, e.g., from foam cells to the liver, is impeded by binding with SR-B1 of oxPCCD36 lipid whiskers protruding from the outer phospholipid shell of oxLDL (see Figures 4 and 5). The consequent accumulation of foam cells in the subendothelial space leads to atherosclerotic plaque formation. Illustration by David Schumick, BS, CMI. Reprinted with permission from Cleveland Clinic Center for Medical Art & Photography. Copyright 2011. All rights reserved.

OxLDL Is a Trojan Horse That Delivers Toxic Electropohilic γ-Hydroxyalkenals into Macrophage and RPE Cells. OxPCCD36-

fostered endocytosis of toxic cargo within an oxLDL particle into CD36-expressing macrophages or RPE cells might lead to (1) lipolysis of oxysterol-containing esters that releases toxic oxysterols or (2) covalent adduction of electrophilic oxidized phospholipids or oxysterols to proteins that impairs protein function. Thus, oxPCCD36 may indirectly promote biological activities such as oxysterol-induced cytotoxicity.38 Covalent adduction of γ-hydroxyalkenal-phospholipids, e.g., HOOA-PC and HODA-PC, with a cysteine thiol in the lysosomal protease cathepsin B, reduces the proteolytic degradation by mouse peritoneal macrophages of macromolecules previously internalized by receptor-mediated endocytosis or phagocytosis.29 Processing of the oxidatively damaged protein may also be impaired by HODA-PC through interference with the fusion with lysosomes of endosomes containing oxLDL in macrophage cells. The analogous processing of PhROS by RPE cells is also perturbed by oxLDL.39 41 It seemed possible that toxic aldehydes present in oxLDL, such as HODA-PC, might inhibit the fusion of endosomes with lysosomes that is crucial for the degradation of oxidatively damaged proteins in endocytosed PhROS. Rab5a is a fusion protein believed to be critical for phagosome and possibly endosome maturation through fusion with lysosomes.42 Indeed, HODA-PC blocked the posttranslational modification, i.e., isoprenylation and proteolytic cleavage, of inactive 25 kDa Rab5a within RPE cells into the active 23 kDa form required for phagosome lysosome fusion in these phagocytes.42 OxPCCD36 Inhibit HDL Binding with Hepatocyte SR-B1, Impeding the Delivery of Cholesterol to the Liver for Excretion. The reverse cholesterol transport pathway is the main mechanism whereby HDL protects against the development of atherosclerosis. It results in the transfer of excess cholesterol from peripheral cells, e.g., macrophages in atherosclerotic plaques, to HDL, which eventually transfers its cholesterol to the

liver for excretion. Efflux of excess cholesterol from macrophages to HDL particles (Figure 8) is followed by conversion of unesterified cholesterol on the particle surface to cholesteryl esters by lecithin/cholesterol acyltransferase (LCAT) and migration to the hydrophobic core. HDL then delivers cholesteryl esters, via the scavenger receptor SR-B1, to the liver where it can be converted into bile acids and excreted. The significant sequence homology and ligands, including HDL, oxLDL, and anionic phospholipids,43 45 between SR-CD36 and SR-B1 suggested that oxPCCD36 might serve as ligands for SR-B1 and thereby interfere with the binding of HDL and reverse cholesterol transport. OxLDL, oxPA-PC, and individual pure oxPCCD36 exhibited saturable binding with human SR-B1.46 Both HDL and oxPCCD36 bound with a purified GST-SR-B1 fusion protein containing the amino acid 144 205 fragment of the extracellular amino-terminal domain of human SR-B1. Importantly, the Kd value of the binding of HDL to this protein was very similar to that determined for small unilammelar vesicles containing oxPCCD36. As expected because of the close proximity of the binding sites for these two ligands on SR-B1, oxLDL and individual pure oxPCCD36 inhibited the binding of HDL with hepatocytes and almost completely inhibited the major physiological function of SR-B1, the selective uptake of cholesteryl esters from HDL by hepatocytes.46 Thus, oxidative stress and accumulation of specific oxidized phospholipids in plasma may promote atherosclerosis not only by inducing the uptake of oxLDL by magrophages via CD36 and interfering with lysosomal processing but also by hampering reverse cholesterol transport by preventing SR-B1-mediated selective uptake of cholesteryl esters into hepatocytes contributing to the development of hypercholesterolemia. OxPCCD36 Induce a Prothrombotic State through the Activation of Platelet CD36. OxPCCD36 also accumulate in the plasma of hyperlipidemic mice at concentrations up to 40fold higher than those found in normolipidemic mice, and they are present in substantial amounts in human plasma at elevated 1796

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

REVIEW

Figure 9. Oxidatively truncated ether phospholipids, referred to collectively as oxPAFs: (A) generation of oxPAFs by oxidative cleavage of PUFA esters of lyso-PAF (LPAF); (B) platelet stimulation by oxPAFs as measured by intracellular Ca2+ as a function of time in platelets treated with 0.5 mmol/L of each phospholipid. Reprinted with permission from ref 50. Copyright 2009 American Heart Association.

levels in individuals with low HDL.47 Through binding with platelet SR-CD36, these levels of oxPCCD36 activate platelets, as assessed by the activation of platelet fibrinogen receptor integrin αIIbβ3 as well as by an increase in P-selectin surface expression.47 This primes or sensitizes the platelets for subsequent activation by the classical platelet agonist ADP. A mesenteric thrombosis model revealed that in vivo occlusion times are significantly shorter in hyperlipidemic mice than in wild-type mice on a Western diet, and functional deficiency of SR-CD36 protects mice from the hyperlipidemia-related prothrombotic phenotype. These observations suggest a role for oxPCCD36 in the pathophysiology of occlusive arterial thrombi associated with myocardial infarction and stroke. They provide a molecular mechanistic link among hyperlipidemia, oxidant stress, and a prothrombotic phenotype. It seems reasonable to anticipate that platelet activation through SR-CD36 promotes thrombosis consequent to the rupture of an atherosclerotic plaque owing to the release of a bolus of oxPCCD36 into the circulation. In normolipidemic human plasma, oxPCCD36 levels are inversely correlated with levels of HDL but not LDL. They are 2.5 times higher in plasma from subjects in the lowest HDL tertile. This is consistent with the anti-inflammatory and antioxidant effects of HDL,48 and suggests that it is an interaction between CD36 and oxidative stress, not merely dyslipidemia (or elevated LDL), which is responsible for enhanced platelet reactivity in vivo. Oxidatively Truncated Ether PCs Activate Platelets through the PAF Receptor. Platelet-activating factor (PAF), 1-O-hexadecyl-2-acetyl-sn-glycero-3-phosphocholine, is an ether phospholipid with potent, diverse physiological actions, particularly as a mediator of inflammation.49 It exerts its effects through a single, highly specific G-protein-coupled receptor. As the most potent phospholipid agonist yet identified, PAF’s biosynthesis is closely controlled. In contrast, PAF-like ether phospholipids with oxidatively truncated sn-2 acyl chains (oxPAFs) can be generated nonenzymatically under oxidative stress. We predicted that the oxPAFs shown in Figure 9A would be generated through the autoxidation of PUFA esters of lyso-PAF (LPAF). To facilitate the identification, quantification, and biological testing of these putative oxPAFs, we prepared pure samples by unambiguous chemical syntheses.50 The electrospray mass spectrum of lipids extracted from oxLDL exhibited molecular ions corresponding to G-LPAF, KOdiA-LPAF, KDdiA-LPAF, KODA-LPAF, and KOOA-

Figure 10. OxPEs exhibit the same action profile as the corresponding oxPCs. Thus, the sn-2 acyl chain, and not the polar headgroup, is the primary determinant of the biological activities tested.

LPAF.50 The entire family of oxidatively truncated ether phospholipids depicted in Figure 9A is generated through oxidative cleavage of 2-lyso-PAF esters of LA, AA, and DHA in small unilamellar vesicles exposed to the biologically relevant myeloperoxidase (MPO)/H2O2/NO2 system to initiate autoxidation. In contrast with the sensitization of platelets toward activation through the binding of oxPC with CD36, oxPAFs are direct platelet agonists that act through the PAF receptor. OxPAFs account for the activation of platelets upon treatment with low concentrations of oxLDL as indicated by the surface expression of P-selectin and the production of highly spread aggregates. A large number of these oxPAFs at submicromolar concentrations induce a rapid increase in platelet cytoplasmic Ca2+ level (Figure 9B), some with higher and some lower activity than 2-lysoPAF (LPAF), but the activity of all oxPAFs and oxLDL is completely suppressed by the PAF receptor antagonist WEB 2086.50 Furthermore, the action profile is bell shaped, with concentrations of oxLDL from just under 1.5 μg/mL up to 5 μg/mL stimulating intracellular Ca2+ flux, but then activation is significantly reduced as the concentration of oxLDL increases above 7 μg/mL, perhaps owing to the presence of antiinflammatory agents in oxLDL. α,β-Unsaturated Carboxylic oxPEs and oxPCs Inhibit LPSInduced Expression of IL-8. OxLDL inhibits the acute 1797

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

REVIEW

Figure 11. Proposed mechanisms for the fragmentation of HOHA-LPAF and the oxidative conversion of HOHA-LPAF into HHdiA-LPAF and KHdiA-LPAF, which are promoted by PUFAs (LH). LO• and LOO• are alkoxy and alkylperoxy radicals, respectively, derived by hydrogen abstraction from LH. LOH, hydroxylipid; MDA, malondialdehyde. Isolable products of these reactions are highlighted. Reprinted with permission from ref 54. Copyright 2008 American Society for Biochemistry and Molecular Biology.

inflammatory response elicited by LPS, a model of bacterial infection, as evidenced by the expression of IL-8 by endothelial cells. Since oxPA-PC and oxPA-PE both mimic this effect, the headgroup is apparently not important for activity. This presumption and the influence of the structure of the oxidatively truncated sn-2 acyl group on biological activity were tested with a variety of analytically pure oxPCs and oxidized phosphatidylethanolamines (oxPEs) available through unambiguous chemical syntheses. Thus, chemical syntheses51 of the oxPEs depicted in Figure 10 enabled the subsequent demonstration of their natural occurrence by LC-MS/MS comparisons with lipids extracted from retina.52 While all of the oxPEs shown in Figure 10 have some activity, those containing α,β-unsaturated carboxylic acids (KHdiA, KOdiA, KDdiA, and HDdiA) are much more potent inhibitors of the LPS induction of IL-8 than are those containing shortchain aldehydes or acids, i.e., OV and G.53 In contrast, the latter are among the most active proinflammatory oxPEs, i.e., for fostering monocyte endothelial interactions. A similar dichotomy is found for oxPCs. Thus, oxidation products with different oxidatively truncated sn-2 acyl chains, as summarized in Figure 10, are most potent in regulating the pro- or anti-inflammatory effects of oxidized phospholipids. The anti-inflammatory inhibition of LPS induction of IL-8 is apparently mediated, at least in part, by the neutral sphingomyelinase since oxPA-PC treatment activates this enzyme and increases levels of the product of its activity, ceramide. Furthermore, the inhibitory activity of oxPA-PC or KOdiA-PC is abrogated by a neutral sphingomyelinase inhibitor, and cell-permeant C6 ceramide inhibits LPS-induced IL-8 synthesis. These effects may reflect ceramideinduced alterations to lipid rafts and caveolae resulting in the deficient assembly of the LPS receptor complex. The noteworthy conclusion is that certain oxPCs can foster the modification of cell membrane structure and thereby alter membrane protein function, e.g., the LPS receptor complex.

’ γ-HYDROXYALKENAL PHOSPHOLIPIDS: BIOLOGICALLY IMPORTANT CHEMISTRY Membrane Lipid Composition Profoundly Influences the Stability of γ-Hydroxy-α,β-unsaturated Aldehydic PCs. Since

phospholipids reside in a membrane environment in vivo, membrane composition is likely to influence their biologically important chemistry. The physiologically relevant MPO/H2O2/ NO2 system can initiate autoxidation of PUFAS by generating •NO2 (see Figure 4) that abstracts doubly allylic hydrogen generating pentadienyl radicals, and these capture oxygen to deliver lipid peroxy (LOO•) and lipid alkoxy (LO•) radicals (Figure 11). Since PUFA-derived LOO• or LO• can oxidize aldehydes and induce fragmentation of α,β-unsaturated aldehydes while •NO2 cannot,54 MPO can only induce these oxidations and fragmentations in membranes that contain PUFAs. We found that PUFAs, e.g., LA-PC in Figure 11, in a membrane promote fragmentation of the ether phospholipid HOHA-LPAF to OB-PAF and oxidation of HOHA-LPAF to HHdiA-LPAF and KHdiALPAF.54 However, these reactions of HOHA-LPAF do not occur readily within membranes composed entirely of saturated diacylPCs. Thus, an abundance of PUFAs in a membrane promotes the removal of γ-hydroxy-α,β-unsaturated aldehydic phospholipids, such as HOHA esters, and thereby disfavors their reactions with proteins to produce biologically active adducts, e.g., carboxyalkylpyrroles (see Figure 2 and the accompanying review17). However, this chemistry does not abolish recognition by SR-CD36 or SR-B1 because products from the oxidation of γ-hydroxy-α,βunsaturated aldehydic oxidized phospholipids, i.e., γ-oxygenatedα,β-unsaturated carboxylic phospholipids, such as HHdiA-PC and KHdiA-PC, retain high affinity for these receptors. Spontaneous Cyclodehydration of HODA-PCs Abolishes Recognition by the Scavenger Receptor CD36. γ-Hydroxyalkenal phospholipids, i.e., HOHA-, HOOA-, and HODA-PCs, 1798

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

REVIEW

Figure 12. Generation of oxPC-furans by cyclodehydration of γ-hydroxyalkenal phospholipids. Reprinted with permission from ref 55. Copyright 2006 American Society for Biochemistry and Molecular Biology.

Figure 13. Summary of the formation and receptor binding of oxPCCD36 and the derived CEP-protein modifications, and their biological sequelae. Binding of the oxidatively truncated γ-hydroxyalkenal PCs, e.g., HOOA-PC, and their more oxidized derivatives, e.g., KOdiA-PC (referred to collectively as oxPCCD36) to the scavenger receptor CD36 activates phagocytic cells or platelets, promoting endocytosis of oxLDL or thrombosis, respectively, while binding with SR-B1 inhibits the interaction with HDL and, thus, interferes with reverse cholesterol transport. The oxidatively truncated γhydroxyalkenal PCs, e.g., HOOA-PC, and their more oxidized derivatives, e.g., KOdiA-PC, readily undergo enzyme-catalyzed phospholipolysis to release lyso-PC. Spontaneous intramolecular transacylation also generates lyso-PC and 5- or 6-membered lactone byproducts. Biologically active shorter chain oxidized PCs can be generated by further fragmentation, e.g., HOOA-PC f OV-PC, while cyclodehydration to oxPC-furans abolishes recognition by CD36. As described in the companion review, covalent adduction of HOHA-PC with proteins in conjunction with phospholipolysis generates a carboxyethylpyrrole (CEP) that activates endothelial cell toll-like receptor (TLR) 2 resulting in migration, proliferation, and tube formation leading to angiogenesis. Illustration by David Schumick, BS, CMI. Reprinted with permission from Cleveland Clinic Center for Medical Art & Photography, Copyright 2011. All rights reserved.

readily undergo another transformation, cyclodehydration, that generates furans, e.g., oxPC-furan(n) where n = the number of methylenes in the alkyl chain (Figure 12).55 This transformation also precludes the reactions of these oxPCs with proteins to produce biologically active adducts, e.g., carboxyalkylpyrroles (see Figure 2 and the accompanying review17), and abolishes recognition by the SR-CD36. Caveat: Lyso-PC Is Generated by Spontaneous Nonenzymatic Deacylation of oxPCs. As noted above, chemical syntheses of pure samples of each oxPCCD36 have been absolutely essential for the unambiguous demonstration of their biological

activities. Although the LC-MS/MS evidence for their presence in lipid mixtures isolated from biological samples is extensive, analytically pure samples of these oxidatively truncated phospholipids have never been isolated from oxPA-PC, oxPL-PC, oxLDL, or human tissues. High purity is crucial when evaluating biological activities because the activities observed could be caused by traces of other components of the original mixture. Furthermore, as noted above, γ-hydroxyalkenals readily undergo free radical-induced spontaneous oxidation to the corresponding γ-ketoalkenals, γ-hydroxyalkenoates, and γ-ketoalkenoates, as well as further oxidative truncation to produce saturated 1799

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology

REVIEW

Figure 14. Generation of lyso-PC from γ- and δ-hydroxy or -keto esters by a nonenzymatic spontaneous transesterification mechanism.

aldehydes, e.g., OB-, OV-, and ON-PC, and carboxylates, e.g., S-, G-, or A-PC, and spontaneous cyclization to oxPC-furans (summarized in Figure 13). Lyso-PC, lacking the sn-2 acyl group, is generated consequent to oxidation of LDL, accounting for nearly one-third56 to one-half57 of the PC equivalents in oxLDL. Elevated levels of lysoPC are linked to cardiovascular complications associated with diabetes, atherosclerosis, and ischemia.58,59 Until recently, it was presumed that lysoPC is produced under physiological conditions by PLA2-mediated hydrolysis of PC60 or from the hydrolysis of oxidized PC by PAF-acetylhydrolase.18,61 However, we recently uncovered yet another process that converts HOHAPC or HOOA-PC into 2-lyso-PC. Notably, this process involves a spontaneous, nonenzymatic intramolecular transesterification mechanism that generates lactone byproducts (Figures 13 and 14). Under physiological conditions, i.e., 37 °C and pH 7.4 in aqueous solution, the half-life for the conversion of HOHA-PC into lysoPC is only 30 min, while that for the conversion of HOOA-PC into lyso-PC is 2 h.62 Furthermore, spontaneous deacylation is not limited to oxPCs that incorporate a hydroxyl group on the γor δ-carbon adjacent to the ester functionality in their oxidized acyl moiety. Oxidatively truncated PCs that incorporate an aldehyde or ketone carbonyl on the γ- or δ-carbon adjacent to the ester functionality also spontaneously deacylate. Thus, KOHA-PC, which has a ketone carbonyl on the γ-carbon adjacent to the ester functionality, spontaneously deacylates with a similar t1/2 ∼ 40 min as the hydroxy analogue HOHA-PC. Presumably, the enedione functional array in KOHA-PC is in rapid equilibrium with a hydrate that cyclizes to generate a hemiacylal with concomitant release of lysoPC (Figure 14). Therefore, it is absolutely essential to ascertain that biological activities attributed to oxPCs that incorporate a hydroxyl or carbonyl group on the γ or δ-carbon adjacent to the ester are not instead activities of lyso-PC. Aqueous solutions of these pure oxPCs, available by chemical syntheses, are especially valuable for the unambiguous demonstration of biological activities. However, such oxPCs are hydrolytically unstable. Their aqueous solutions must be freshly prepared and kept cold until use.

’ CONCLUSIONS One approach to identify oxidized phospholipids formed in vivo is to predict likely candidates by mechanistic speculation or analogy with known products of lipid oxidation and to use

authentic samples of the putative natural products, prepared by unambiguous chemical synthesis, to guide and confirm LC-MS/ MS analyses of biological extracts. Using this approach, the formation in vivo of families of diacyl oxPEs and oxPCs, as well as sn-1-alkyl-sn-2-acyl oxPCs containing truncated sn-2 acyl groups with γ-hydroxyalkenal or more oxidized functionalities, were established, and several pathways for their spontaneous decomposition, including the generation of 2-lyso-PC, were discovered. Their involvement as ligands (Figure 13) for receptor-mediated endocytosis of oxLDL by macrophage cells and of oxidatively damaged rod photoreceptor cells by retinal pigmented endothelial cells, blockage of reverse cholesterol transport to the liver, and sensitization or activation of platelets toward thrombosis were demonstrated, as were other involvements in chronic inflammation and inhibition of normal processing of endocytosed oxLDL, e.g., through covalent protein modification that causes a loss of function of proteases and proteins that enable phagosome and possibly endosome maturation through fusion with lysosomes. Covalent γ-hydroxyalkenal-PC-derived protein modifications that result in a gain of function are the focus of the accompanying review.17

’ AUTHOR INFORMATION Corresponding Author

*Phone: 216-368-2592. Fax: 216-368-3006. E-mail: [email protected]. Funding Sources

We are grateful for the financial support by National Institutes of Health Grants GM021249, HL53315, and EY016813 for studies in the laboratory of R.G.S. presented in this review.

’ ACKNOWLEDGMENT We thank David Schumick for creating Figures 4, 6, 8, and 13. ’ ABBREVIATIONS CHP, carboxyheptyl pyrrole; CPP, carboxypropyl pyrrole; G-PC, 1palmityl-2-glutaryl-sn-glycero-3-phosphocholine; HDL, high-density lipoprotein; HNE, γ-hydroxyalkenal 4-hydroxy-2-nonenal; 13-HODE-PC, 13-hydroxyoctadeca-9,11-dienoyl phosphatidylcholine; HOOA-PC, 1-palmityl-2-(5-hydroxy-8-oxooct-6-enoyl)-sn-glycero3-phosphocholine; IL-8, interleukin-8; KOdiA-PC, 5-ketooct-6endioyl phosphatidylcholine; LCAT, lecithin/cholesterol acyltransferase; LDL, low-density lipoprotein; LPAF, lyso-PAF 1800

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology (1-O-hexadecyl-2-lyso-sn-glycero-3-phosphocholine); LPS, lipopolysaccharide; moxLDL, minimally oxidized low-density lipoprotein; MPO, myeloperoxidase; OV-PC, 1-palmityl-2-(ωoxovaleryl)-sn-glycero-3-phosphocholine; oxLDL, oxidized lowdensity lipoprotein; oxPC, oxidized phosphatidylcholine; oxPE, oxidized phosphatidylethanolamine; PAF, platelet-activating factor (1-O-hexadecyl-2-acetyl-sn-glycero-3-phosphocholine); PA-PC, 1palmityl-2-arachidonoyl-sn-glycero-3-phosphocholine; PhROS, photoreceptor rod cell outer segments; PL-PC, 1-palmityl-2-linoleyl-sn-glycero-3-phosphocholine; PUFA, polyunsaturated fatty acid; RPE, retinal pigmented epithelial; SR, scavenger receptor.

’ REFERENCES (1) Berliner, J. A., Subbanagounder, G., Leitinger, N., Watson, A. D., and Vora, D. (2001) Evidence for a role of phospholipid oxidation products in atherogenesis. Trends Cardiovasc. Med. 11, 142–147. (2) Davies, S. S., Pontsler, A. V., Marathe, G. K., Harrison, K. A., Murphy, R. C., Hinshaw, J. C., Prestwich, G. D., Hilaire, A. S., Prescott, S. M., Zimmerman, G. A., and McIntyre, T. M. (2001) Oxidized alkyl phospholipids are specific, high affinity peroxisome proliferator-activated receptor gamma ligands and agonists. J. Biol. Chem. 276, 16015–16023. (3) Kayganich, K. A., and Murphy, R. C. (1992) Fast atom bombardment tandem mass spectrometric identification of diacyl, alkylacyl, and alk-1-enylacyl molecular species of glycerophosphoethanolamine in human polymorphonuclear leukocytes. Anal. Chem. 64, 2965–2971. (4) Kogure, K., Nakashima, S., Tsuchie, A., Tokumura, A., and Fukuzawa, K. (2003) Temporary membrane distortion of vascular smooth muscle cells is responsible for their apoptosis induced by platelet-activating factor-like oxidized phospholipids and their degradation product, lysophosphatidylcholine. Chem. Phys. Lipids 126, 29–38. (5) Marathe, G. K., Davies, S. S., Harrison, K. A., Silva, A. R., Murphy, R. C., Castro-Faria-Neto, H., Prescott, S. M., Zimmerman, G. A., and McIntyre, T. M. (1999) Inflammatory platelet-activating factor-like phospholipids in oxidized low density lipoproteins are fragmented alkyl phosphatidylcholines. J. Biol. Chem. 274, 28395–28404. (6) McIntyre, T. M., Zimmerman, G. A., and Prescott, S. M. (1999) Biologically active oxidized phospholipids. J. Biol. Chem. 274, 25189– 25192. (7) Nakamura, T., Bratton, D. L., and Murphy, R. C. (1997) Analysis of epoxyeicosatrienoic and monohydroxyeicosatetraenoic acids esterified to phospholipids in human red blood cells by electrospray tandem mass spectrometry. J. Mass Spectrom. 32, 888–896. (8) Tokumura, A., Fukuzawa, K., Akamatsu, Y., Yamada, S., Suzuki, T., and Tsukatani, H. (1978) Identification of vasopressor phospholipid in crude soybean lecithin. Lipids 13, 468–472. (9) Tokumura, A., Sumida, T., Toujima, M., Kogure, K., and Fukuzawa, K. (2000) Platelet-activating factor (PAF)-like oxidized phospholipids: relevance to atherosclerosis. BioFactors (Oxford, England) 13, 29–33. (10) Tokumura, A., Sumida, T., Toujima, M., Kogure, K., Fukuzawa, K., Takahashi, Y., and Yamamoto, S. (2000) Structural identification of phosphatidylcholines having an oxidatively shortened linoleate residue generated through its oxygenation with soybean or rabbit reticulocyte lipoxygenase. J. Lipid Res. 41, 953–962. (11) Tokumura, A., Toujima, M., Yoshioka, Y., and Fukuzawa, K. (1996) Lipid peroxidation in low density lipoproteins from human plasma and egg yolk promotes accumulation of 1-acyl analogues of platelet-activating factor-like lipids. Lipids 31, 1251–1258. (12) Berliner, J. A., and Watson, A. D. (2005) A role for oxidized phospholipids in atherosclerosis. New Engl. J. Med. 353, 9–11. (13) McIntyre, T. M., and Hazen, S. L. (2010) Lipid oxidation and cardiovascular disease: introduction to a review series. Circ. Res. 107, 1167–1169. (14) Spiteller, G. (2005) The relation of lipid peroxidation processes with atherogenesis: a new theory on atherogenesis. Mol. Nutr. Food Res. 49, 999–1013.

REVIEW

(15) Witztum, J. L., and Berliner, J. A. (1998) Oxidized phospholipids and isoprostanes in atherosclerosis. Curr. Opin. Lipidol. 9, 441– 448. (16) Horiguchi, T., Rithner, C. D., Croteau, R., and Williams, R. M. (2003) Studies on taxol biosynthesis. Preparation of 5α-acetoxytaxa4(20),11-dien-2 α,10β-diol derivatives by deoxygenation of a taxadiene tetra-acetate obtained from Japanese yew. Tetrahedron 59, 267–273. (17) Salomon, R. G., Hong, L., and Hollyfield, J. G. (2011) Discovery of carboxyethylpyrroles (CEPs): critical insights into AMD, autism, cancer, and wound healing from basic research on the chemistry of oxidized phospholipids. Chem. Res. Toxicol., DOI: 10.1021/ tx200206v. (18) Stremler, K. E., Stafforini, D. M., Prescott, S. M., Zimmerman, G. A., and McIntyre, T. M. (1989) An oxidized derivative of phosphatidylcholine is a substrate for the platelet-activating factor acetylhydrolase from human plasma. J. Biol. Chem. 264, 5331–5334. (19) Watson, A. D., Leitinger, N., Navab, M., Faull, K. F., Horkko, S., Witztum, J. L., Palinski, W., Schwenke, D., Salomon, R. G., Sha, W., Subbanagounder, G., Fogelman, A. M., and Berliner, J. A. (1997) Structural identification by mass spectrometry of oxidized phospholipids in minimally oxidized low density lipoprotein that induce monocyte/ endothelial interactions and evidence for their presence in vivo. J. Biol. Chem. 272, 13597–13607. (20) Esterbauer, H., Schaur, R. J., and Zollner, H. (1991) Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radical Biol. Med. 11, 81–128. (21) Sayre, L. M., Sha, W., Xu, G., Kaur, K., Nadkarni, D., Subbanagounder, G., and Salomon, R. G. (1996) Immunochemical evidence supporting 2-pentylpyrrole formation on proteins exposed to 4-hydroxy-2-nonenal. Chem. Res. Toxicol. 9, 1194–1201. (22) Kaur, K., Salomon, R. G., O’Neil, J., and Hoff, H. F. (1997) (Carboxyalkyl)pyrroles in human plasma and oxidized low-density lipoproteins. Chem. Res. Toxicol. 10, 1387–1396. (23) Salomon, R. G., Batyreva, E., Kaur, K., Sprecher, D. L., Schreiber, M. J., Crabb, J. W., Penn, M. S., DiCorletoe, A. M., Hazen, S. L., and Podrez, E. A. (2000) Isolevuglandin-protein adducts in humans: products of free radical-induced lipid oxidation through the isoprostane pathway. Biochim. Biophys. Acta 1485, 225–235. (24) Berg, J. M., Tymoczko, J. L., and Stryer, L. (2007) Biochemistry, in Biochemistry, p 67, W. H. Freeman, New York. (25) Spiteller, P., Kern, W., Reiner, J., and Spiteller, G. (2001) Aldehydic lipid peroxidation products derived from linoleic acid. Biochim. Biophys. Acta 1531, 188–208. (26) Deng, Y., and Salomon, R. G. (1998) Total synthesis of γ-hydroxy-α,β-unsaturated aldehydic esters of cholesterol and 2-lysophosphatidylcholine. J. Org. Chem. 63, 7789–7794. (27) Subbanagounder, G., Deng, Y., Borromeo, C., Dooley, A. N., Berliner, J. A., and Salomon, R. G. (2002) Hydroxy alkenal phospholipids regulate inflammatory functions of endothelial cells. Vasc. Pharmacol. 38, 201–209. (28) Podrez, E. A., Poliakov, E., Shen, Z., Zhang, R., Deng, Y., Sun, M., Finton, P. J., Shan, L., Febbraio, M., Hajjar, D. P., Silverstein, R. L., Hoff, H. F., Salomon, R. G., and Hazen, S. L. (2002) A novel family of atherogenic oxidized phospholipids promotes macrophage foam cell formation via the scavenger receptor CD36 and is enriched in atherosclerotic lesions. J. Biol. Chem. 277, 38517–38523. (29) Hoff, H. F., O’Neil, J., Wu, Z., Hoppe, G., and Salomon, R. L. (2003) Phospholipid hydroxyalkenals: biological and chemical properties of specific oxidized lipids present in atherosclerotic lesions. Arterioscler., Thromb., Vasc. Biol. 23, 275–282. (30) Zhang, R., Brennan, M. L., Fu, X., Aviles, R. J., Pearce, G. L., Penn, M. S., Topol, E. J., Sprecher, D. L., and Hazen, S. L. (2001) Association between myeloperoxidase levels and risk of coronary artery disease. JAMA 286, 2136–2142. (31) Li, X. M., Salomon, R. G., Qin, J., and Hazen, S. L. (2007) Conformation of an endogenous ligand in a membrane bilayer for the macrophage scavenger receptor CD36. Biochemistry 46, 5009– 5017. 1801

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802

Chemical Research in Toxicology (32) Greenberg, M. E., Li, X. M., Gugiu, B. G., Gu, X., Qin, J., Salomon, R. G., and Hazen, S. L. (2008) The lipid whisker model of the structure of oxidized cell membranes. J. Biol. Chem. 283, 2385–2396. (33) Khandelia, H., and Mouritsen, O. G. (2009) Lipid gymnastics: evidence of complete acyl chain reversal in oxidized phospholipids from molecular simulations. Biophys. J. 96, 2734–2743. (34) Epand, R. F., Mishra, V. K., Palgunachari, M. N., Anantharamaiah, G. M., and Epand, R. M. (2009) Anti-inflammatory peptides grab on to the whiskers of atherogenic oxidized lipids. Biochim. Biophys. Acta 1788, 1967–1975. (35) Podrez, E. A., Poliakov, E., Shen, Z., Zhang, R., Deng, Y., Sun, M., Finton, P. J., Shan, L., Gugiu, B., Fox, P. L., Hoff, H. F., Salomon, R. G., and Hazen, S. L. (2002) Identification of a novel family of oxidized phospholipids that serve as ligands for the macrophage scavenger receptor CD36. J. Biol. Chem. 277, 38503–38516. (36) Sun, M., Deng, Y., Batyreva, E., Sha, W., and Salomon, R. G. (2002) Novel bioactive phospholipids: practical total syntheses of products from the oxidation of arachidonic and linoleic esters of 2-lysophosphatidylcholine(1). J. Biol. Chem. 67, 3575–3584. (37) Sun, M., Finnemann, S. C., Febbraio, M., Shan, L., Annangudi, S. P., Podrez, E. A., Hoppe, G., Darrow, R., Organisciak, D. T., Salomon, R. G., Silverstein, R. L., and Hazen, S. L. (2006) Light-induced oxidation of photoreceptor outer segment phospholipids generates ligands for CD36-mediated phagocytosis by retinal pigment epithelium: a potential mechanism for modulating outer segment phagocytosis under oxidant stress conditions. J. Biol. Chem. 281, 4222–4230. (38) Colles, S. M., Irwin, K. C., and Chisolm, G. M. (1996) Roles of multiple oxidized LDL lipids in cellular injury: dominance of 7 betahydroperoxycholesterol. J. Lipid Res. 37, 2018–2028. (39) Hoppe, G., Marmorstein, A. D., Pennock, E. A., and Hoff, H. F. (2001) Oxidized low density lipoprotein-induced inhibition of processing of photoreceptor outer segments by RPE. Invest. Ophthalmol. Vis. Sci. 42, 2714–2720. (40) Hoppe, G., O’Neil, J., Hoff, H. F., and Sears, J. (2004) Accumulation of oxidized lipid-protein complexes alters phagosome maturation in retinal pigment epithelium. Cell. Mol. Life Sci. 61, 1664– 1674. (41) Hoppe, G., O’Neil, J., Hoff, H. F., and Sears, J. (2004) Products of lipid peroxidation induce missorting of the principal lysosomal protease in retinal pigment epithelium. Biochim. Biophys. Acta 1689, 33–41. (42) Alvarez-Dominguez, C., and Stahl, P. D. (1998) Interferongamma selectively induces Rab5a synthesis and processing in mononuclear cells. J. Biol. Chem. 273, 33901–33904. (43) Connelly, M. A., and Williams, D. L. (2004) Scavenger receptor BI: a scavenger receptor with a mission to transport high density lipoprotein lipids. Curr. Opin. Lipidol. 15, 287–295. (44) Jessup, W., Gelissen, I. C., Gaus, K., and Kritharides, L. (2006) Roles of ATP binding cassette transporters A1 and G1, scavenger receptor BI and membrane lipid domains in cholesterol export from macrophages. Curr. Opin. Lipidol. 17, 247–257. (45) Rigotti, A., Miettinen, H. E., and Krieger, M. (2003) The role of the high-density lipoprotein receptor SR-BI in the lipid metabolism of endocrine and other tissues. Endocr. Rev. 24, 357–387. (46) Ashraf, M. Z., Kar, N. S., Chen, X., Choi, J., Salomon, R. G., Febbraio, M., and Podrez, E. A. (2008) Specific oxidized phospholipids inhibit scavenger receptor bi-mediated selective uptake of cholesteryl esters. J. Biol. Chem. 283, 10408–10414. (47) Podrez, E. A., Byzova, T. V., Febbraio, M., Salomon, R. G., Ma, Y., Valiyaveettil, M., Poliakov, E., Sun, M., Finton, P. J., Curtis, B. R., Chen, J., Zhang, R., Silverstein, R. L., and Hazen, S. L. (2007) Platelet CD36 links hyperlipidemia, oxidant stress and a prothrombotic phenotype. Nature Med. 13, 1086–1095. (48) Barter, P. J., Nicholls, S., Rye, K. A., Anantharamaiah, G. M., Navab, M., and Fogelman, A. M. (2004) Antiinflammatory properties of HDL. Circ. Res. 95, 764–772. (49) Prescott, S. M., Zimmerman, G. A., Stafforini, D. M., and McIntyre, T. M. (2000) Platelet-activating factor and related lipid mediators. Annu. Rev. Biochem. 69, 419–445.

REVIEW

(50) Chen, R., Chen, X., Salomon, R. G., and McIntyre, T. M. (2009) Platelet activation by low concentrations of intact oxidized LDL particles involves the PAF receptor. Arterioscler., Thromb., Vasc., Biol. 29, 363–371. (51) Gugiu, B. G., and Salomon, R. G. (2003) Total syntheses of bioactive oxidized ethanolamine phospholipids. Org. Lett. 5, 2797–2799. (52) Gugiu, B. G., Mesaros, C. A., Sun, M., Gu, X., Crabb, J. W., and Salomon, R. G. (2006) Identification of oxidatively truncated ethanolamine phospholipids in retina and their generation from polyunsaturated phosphatidylethanolamines. Chem. Res. Toxicol. 19, 262–271. (53) Walton, K. A., Gugiu, B. G., Thomas, M., Basseri, R. J., Eliav, D. R., Salomon, R. G., and Berliner, J. A. (2006) A role for neutral sphingomyelinase activation in the inhibition of LPS action by phospholipid oxidation products. J. Lipid Res. 47, 1967–1974. (54) Chen, X., Zhang, W., Laird, J., Hazen, S. L., and Salomon, R. G. (2008) Polyunsaturated phospholipids promote the oxidation and fragmentation of gamma-hydroxyalkenals: formation and reactions of oxidatively truncated ether phospholipids. J. Lipid Res. 49, 832–846. (55) Gao, S., Zhang, R., Greenberg, M. E., Sun, M., Chen, X., Levison, B. S., Salomon, R. G., and Hazen, S. L. (2006) Phospholipid hydroxyalkenals, a subset of recently discovered endogenous CD36 ligands, spontaneously generate novel furan-containing phospholipids lacking CD36 binding activity in vivo. J. Biol. Chem. 281, 31298–31308. (56) Steinbrecher, U. P., Zhang, H. F., and Lougheed, M. (1990) Role of oxidatively modified LDL in atherosclerosis. Free Radical Biol. Med. 9, 155–168. (57) Sakai, M., Miyazaki, A., Hakamata, H., Sasaki, T., Yui, S., Yamazaki, M., Shichiri, M., and Horiuchi, S. (1994) Lysophosphatidylcholine plays an essential role in the mitogenic effect of oxidized low density lipoprotein on murine macrophages. J. Biol. Chem. 269, 31430– 31435. (58) Shi, A., Yoshinari, M., Iino, K., Wakisaka, M., Iwase, M., and Fujishima, M. (1999) Lysophosphatidylcholine molecular species in low density lipoprotein and high density lipoprotein in alloxan-induced diabetic rats: effect of probucol. Exp. Clin. Endocrinol. Diabetes 107, 337–342. (59) Shi, A. H., Yoshinari, M., Wakisaka, M., Iwase, M., and Fujishima, M. (1999) Lysophosphatidylcholine molecular species in low density lipoprotein of type 2 diabetes. Horm. Metab. Res. 31, 283–286. (60) Parthasarathy, S., and Barnett, J. (1990) Phospholipase A2 activity of low density lipoprotein: evidence for an intrinsic phospholipase A2 activity of apoprotein B-100. Proc. Natl. Acad. Sci. U.S.A. 87, 9741–9745. (61) Tew, D. G., Southan, C., Rice, S. Q., Lawrence, M. P., Li, H., Boyd, H. F., Moores, K., Gloger, I. S., and Macphee, C. H. (1996) Purification, properties, sequencing, and cloning of a lipoprotein-associated, serine-dependent phospholipase involved in the oxidative modification of low-density lipoproteins. Arterioscler., Thromb., Vasc. Biol. 16, 591–599. (62) Choi, J., Zhang, W., Gu, X., Chen, X., Hong, L., Laird, J. M., and Salomon, R. G. (2011) Lysophosphatidylcholine is generated by spontaneous deacylation of oxidized phospholipids. Chem. Res. Toxicol. 24, 111–118.

1802

dx.doi.org/10.1021/tx200207z |Chem. Res. Toxicol. 2011, 24, 1791–1802